Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Gladys O. Latunde-Dada is active.

Publication


Featured researches published by Gladys O. Latunde-Dada.


Cell | 2005

Identification of an Intestinal Heme Transporter

M Shayeghi; Gladys O. Latunde-Dada; Jonathan S. Oakhill; Abas H. Laftah; Ken Takeuchi; Neil Halliday; Yasmin Khan; Alice Warley; Fiona E. McCann; Robert C. Hider; David M. Frazer; Gregory J. Anderson; Chris D. Vulpe; Robert J. Simpson; Andrew T. McKie

Dietary heme iron is an important nutritional source of iron in carnivores and omnivores that is more readily absorbed than non-heme iron derived from vegetables and grain. Most heme is absorbed in the proximal intestine, with absorptive capacity decreasing distally. We utilized a subtractive hybridization approach to isolate a heme transporter from duodenum by taking advantage of the intestinal gradient for heme absorption. Here we show a membrane protein named HCP 1 (heme carrier protein 1), with homology to bacterial metal-tetracycline transporters, mediates heme uptake by cells in a temperature-dependent and saturable manner. HCP 1 mRNA was highly expressed in duodenum and regulated by hypoxia. HCP 1 protein was iron regulated and localized to the brush-border membrane of duodenal enterocytes in iron deficiency. Our data indicate that HCP 1 is the long-sought intestinal heme transporter.


FEBS Letters | 2006

Haem carrier protein 1 (HCP1): Expression and functional studies in cultured cells

Gladys O. Latunde-Dada; Ken Takeuchi; Robert J. Simpson; Andrew T. McKie

Haem released from digestion and breakdown of meat products provides an important source of dietary iron, which is readily absorbed in the proximal intestine. The recent cloning and characterization of a haem carrier protein 1 (HCP 1) has provided a candidate intestinal haem transporter. The current studies describe the expression and functional analysis of HCP1 in cultured Caco‐2 cells, a commonly used model of human intestinal cells. HCP1 mRNA expression in other cell types was also studied. The uptake of 55Fe labeled haem was determined in cells under different experimental conditions and HCP1 expression was measured by RT‐PCR and immunohistochemistry. mRNA and protein expressions increased in Caco‐2 cells transduced with HCP1 adenoviral plasmid, and consequently 55Fe haem uptake was higher in these cells. Haem uptake was also increased in fully differentiated Caco‐2 cells compared to undifferentiated cells. Preincubation of cells with desferrioxamine (DFO, to deplete cells of iron) had no effect on HCP1 expression or haem uptake. Treatment with CdCl2 (to induce haem oxygenase, HO‐1) enhanced HCP1 expression and increased haem uptake into the cells. HCP1 expression and function were found to be adaptive to the rate of haem degradation by HO‐1. Furthermore, HCP1 expression in different cells implies a functional role in tissues other than the duodenum.


Blood | 2009

Identification of a Steap3 endosomal targeting motif essential for normal iron metabolism

Teresa Lambe; Robert J. Simpson; Sara Dawson; Tiphaine Bouriez-Jones; Tanya L. Crockford; Michelle Lepherd; Gladys O. Latunde-Dada; Hannah Robinson; Kishor B. Raja; Dean R. Campagna; Guadalupe Villarreal; J. Clive Ellory; Christopher C. Goodnow; Mark D. Fleming; Andrew T. McKie; Richard J. Cornall

Hereditary forms of iron-deficiency anemia, including animal models, have taught us much about the normal physiologic control of iron metabolism. However, the discovery of new informative mutants is limited by the natural mutation frequency. To address this limitation, we have developed a screen for heritable abnormalities of red blood cell morphology in mice with single-nucleotide changes induced by the chemical mutagen ethylnitrosourea (ENU). We now describe the first strain, fragile-red, with hypochromic microcytic anemia resulting from a Y228H substitution in the ferrireductase Steap3 (Steap3(Y288H)). Analysis of the Steap3(Y288H) mutant identifies a conserved motif required for targeting Steap3 to internal compartments and highlights how phenotypic screens linked to mutagenesis can identify new functional variants in erythropoiesis and ascribe function to previously unidentified motifs.


British Journal of Nutrition | 2009

Haem and folate transport by proton-coupled folate transporter/haem carrier protein 1 (SLC46A1)

Abas H. Laftah; Gladys O. Latunde-Dada; Sarah Fakih; Robert C. Hider; Robert J. Simpson; Andrew T. McKie

Haem carrier protein 1 (HCP1) was originally identified and characterised as a mammalian haem transporter. However, recent evidence has shown that it is also a proton-coupled folate transporter (PCFT) and mutations in the gene cause hereditary folate deficiency in humans. We therefore investigated haem and folate transport characteristics of PCFT/HCP1 both in vivo and in vitro in CD-1 mice and in the presence or absence of a blocking antibody for PCFT/HCP1, and also in cultured cells (which express PCFT/HCP1 endogenously) to elucidate the specificity and selectivity of PCFT/HCP1. The in vivo study showed that the addition of folic acid inhibited 59Fe-labelled haem transport in hypoxic mice but had no effect in normal mice. Using in vitro methods, the results showed increased [3H]folate uptake into everted duodenum from hypoxic mice but uptake was reduced by the addition of haem or PCFT/HCP1 antibodies to the medium. Caco-2 cells transiently transfected with small interfering RNA (siRNA) PCFT/HCP1 duplex oligos resulted in a 69 % reduction in PCFT/HCP1 mRNA when compared with the control siRNA. Both haem and folate uptake were significantly (P < 0.05) reduced in cells transfected with PCFT/HCP1 siRNA; however, the magnitude of reduction with folic acid uptake was greater (48 %) than that of haem (22.5 %). Overall the data support PCFT/HCP1 as a primary folate transporter with a lower affinity for haem. PCFT/HCP1 could therefore play a physiological role in Fe nutrition and the data highlight the potential for the interaction of folate and haem at the level of intestinal absorption.


Scandinavian Journal of Gastroenterology | 2005

Expression of iron absorption genes in mouse large intestine

Ken Takeuchi; Ingvar Bjarnason; Abas H. Laftah; Gladys O. Latunde-Dada; Robert J. Simpson; Andrew T. McKie

Objective The large intestine has been reported to have a capacity for iron absorption and expresses genes for iron absorption normally found in the duodenum. The importance and function of these genes in the large intestine are not understood. We therefore investigated the cellular localization and regulation of expression of these genes in mouse caecum and colon. Material and methods Gene expression was measured by real-time PCR using RNA extracted from iron-deficient and hypoxic mouse large intestine, compared to controls. Protein localization and regulation were measured by immunohistochemistry using frozen sections of the large intestine from the same mice. Results Dcytb (duodenal ferric reductase) was expressed at very low levels in the large intestine, compared to the duodenum, while Ireg1 and DMT1 were expressed at significant levels in the large intestine and were increased in iron-deficient caecum, proximal and distal colon, with the most significant increases seen in the distal colon. Hypoxia increased Ireg1 expression in the proximal colon. Immunohistochemistry detected significant levels of only IREG1, which was localized to the basolateral membrane of colonic epithelial cells. Conclusions Iron absorption genes were expressed at lower levels in mouse caecum and colon than in the duodenum. They are regulated by body iron requirements. Colonic epithelial cells express basolateral IREG1in the same fashion as in the duodenum and this protein could regulate colonic epithelial cell iron levels.


Nanomedicine: Nanotechnology, Biology and Medicine | 2014

A nano-disperse ferritin-core mimetic that efficiently corrects anemia without luminal iron redox activity

Jonathan J. Powell; Sylvaine F. A. Bruggraber; Nuno Faria; Lynsey K. Poots; Nicole Hondow; Timothy J. Pennycook; Gladys O. Latunde-Dada; Robert J. Simpson; Andy Brown; Dora I. A. Pereira

The 2-5 nm Fe(III) oxo-hydroxide core of ferritin is less ordered and readily bioavailable compared to its pure synthetic analogue, ferrihydrite. We report the facile synthesis of tartrate-modified, nano-disperse ferrihydrite of small primary particle size, but with enlarged or strained lattice structure (~ 2.7 Å for the main Bragg peak versus 2.6 Å for synthetic ferrihydrite). Analysis indicated that co-precipitation conditions can be achieved for tartrate inclusion into the developing ferrihydrite particles, retarding both growth and crystallization and favoring stabilization of the cross-linked polymeric structure. In murine models, gastrointestinal uptake was independent of luminal Fe(III) reduction to Fe(II) and, yet, absorption was equivalent to that of ferrous sulphate, efficiently correcting the induced anemia. This process may model dietary Fe(III) absorption and potentially provide a side effect-free form of cheap supplemental iron. From the Clinical Editor Small size tartrate-modified, nano-disperse ferrihydrite was used for efficient gastrointestinal delivery of soluble Fe(III) without the risk for free radical generation in murine models. This method may provide a potentially side effect-free form iron supplementation.


Journal of Nutrition | 2012

Duodenal Reductase Activity and Spleen Iron Stores Are Reduced and Erythropoiesis Is Abnormal in Dcytb Knockout Mice Exposed to Hypoxic Conditions

Jeehyea Choi; Patarabutr Masaratana; Gladys O. Latunde-Dada; Matthew Arno; Robert J. Simpson; Andrew T. McKie

Duodenal cytochrome b (Dcytb, Cybrd1) is a ferric reductase localized in the duodenum that is highly upregulated in circumstances of increased iron absorption. To address the contribution of Dcytb to total duodenal ferric reductase activity as well as its wider role in iron metabolism, we first measured duodenal ferric reductase activity in wild-type (WT) and Dcytb knockout (Dcytb(-/-)) mice under 3 conditions known to induce gut ferric reductase: dietary iron deficiency, hypoxia, and pregnancy. Dcytb(-/-) and WT mice were randomly assigned to control (iron deficiency experiment, 48 mg/kg dietary iron; hypoxia experiment, normal atmospheric pressure; pregnancy experiment, nonpregnant animals) or treatment (iron deficiency experiment, 2-3 mg/kg dietary iron; hypoxia experiment, 53.3 kPa pressure; pregnancy experiment, d 20 of pregnancy) groups and duodenal reductase activity measured. We found no induction of ferric reductase activity in Dcytb(-/-) mice under any of these conditions, indicating there are no other inducible ferric reductases present in the duodenum. To test whether Dcytb was required for iron absorption in conditions with increased erythropoietic demand, we also measured tissue nonheme iron levels and hematological indices in WT and Dcytb(-/-) mice exposed to hypoxia. There was no evidence of gross alterations in iron absorption, hemoglobin, or total liver nonheme iron in Dcytb(-/-) mice exposed to hypoxia compared with WT mice. However, spleen nonheme iron was significantly less (6.7 ± 1.0 vs. 12.7 ± 0.9 nmol · mg tissue(-1); P < 0.01, n = 7-8) in hypoxic Dcytb(-/-) compared with hypoxic WT mice and there was evidence of impaired reticulocyte hemoglobinization with a lower reticulocyte mean corpuscular hemoglobin (276 ± 1 vs. 283 ± 2 g · L(-1); P < 0.05, n = 7-8) in normoxic Dcytb(-/-) compared with normoxic WT mice. We therefore conclude that DCYTB is the primary iron-regulated duodenal ferric reductase in the gut and that Dcytb is necessary for optimal iron metabolism.


Journal of Biological Chemistry | 2012

BMPER Protein Is a Negative Regulator of Hepcidin and Is Up-regulated in Hypotransferrinemic Mice

Neeta Patel; Patarabutr Masaratana; Javier Díaz-Castro; Gladys O. Latunde-Dada; Aakafa Qureshi; Pamela Lockyer; Molly Jacob; Matthew Arno; Pavle Matak; Ragai R. Mitry; Robin D. Hughes; Anil Dhawan; Cam Patterson; Robert J. Simpson; Andrew T. McKie

Background: The mechanism by which anemia results in lowered hepcidin levels is not clear. Results: Bone morphogenetic protein (BMP)-binding endothelial cell precursor-derived regulator (BMPER), a known BMP antagonist, was found to be up-regulated in anemic Trfhpx/hpx mice and to suppress hepcidin transcription both in vivo and in vitro. Conclusion: BMPER is involved in suppressing hepcidin levels in Trfhpx/hpx mice. Significance: BMPER is a novel regulator of hepcidin and iron metabolism. The BMP/SMAD4 pathway has major effects on liver hepcidin levels. Bone morphogenetic protein-binding endothelial cell precursor-derived regulator (Bmper), a known regulator of BMP signaling, was found to be overexpressed at the mRNA and protein levels in liver of genetically hypotransferrinemic mice (Trfhpx/hpx). Soluble BMPER peptide inhibited BMP2- and BMP6-dependent hepcidin promoter activity in both HepG2 and HuH7 cells. These effects correlated with reduced cellular levels of pSMAD1/5/8. Addition of BMPER peptide to primary human hepatocytes abolished the BMP2-dependent increase in hepcidin mRNA, whereas injection of Bmper peptide into mice resulted in reduced liver hepcidin and increased serum iron levels. Thus Bmper may play an important role in suppressing hepcidin production in hypotransferrinemic mice.


Biochimica et Biophysica Acta | 2017

Ferroptosis: Role of lipid peroxidation, iron and ferritinophagy

Gladys O. Latunde-Dada

Ferroptosis is a form of regulated cell death that is dependent on iron and reactive oxygen species (ROS) and is characterized by lipid peroxidation. It is morphologically and biochemically distinct and disparate from other processes of cell death. As ferroptosis is induced by inhibition of cysteine uptake or inactivation of the lipid repair enzyme glutathione peroxidase 4 (GPX4), the process is favored by chemical or mutational inhibition of the cystine/glutamate antiporter and culminates in the accumulation of reactive oxygen species (ROS) in the form of lipid hydroperoxides. Excessive lipid peroxidation leads to death by ferroptosis and the phenotype is accentuated respectively by the repletion and depletion of iron and glutathione in cells. Furthermore, oxidized phosphatidylethanolamines (PE) harbouring arachidonoyl (AA) and adrenoyl moieties (AdA) have been shown as proximate executioners of ferroptosis. Induction of ferroptosis due to cysteine depletion leads to the degradation of ferritin (i.e. ferritinophagy), which releases iron via the NCOA4-mediated autophagy pathway. Evidence of the manifestation of ferroptosis in vivo in iron overload mice mutants is emerging. Thus, a concerted synchronization of iron availability, ROS generation, glutamate excess and cysteine deficit leads to ferroptosis. A number of questions on the molecular mechanisms of some features of ferroptosis are highlighted as subjects for future investigations.


BioEssays | 2009

Iron metabolism: microbes, mouse, and man.

Gladys O. Latunde-Dada

Recent advances in research on iron metabolism have revealed the identity of a number of genes, signal transduction pathways, and proteins involved in iron regulation in mammals. The emerging paradigm is a coordination of homeostasis within a network of classical iron metabolic pathways and other cellular processes such as cell differentiation, growth, inflammation, immunity, and a host of physiologic and pathologic conditions. Iron, immunity, and infection are intricately linked and their regulation is fundamental to the survival of mammals. The mutual dependence on iron by the host and invading pathogenic organisms elicits competition for the element during infection. While the host maintains mechanisms to utilize iron for its own metabolism exclusively, pathogenic organisms are armed with a myriad of strategies to circumvent these measures. This review explores iron metabolism in mammalian host, defense mechanisms against pathogenic microbes and the competitive devices of microbes for access to iron.

Collaboration


Dive into the Gladys O. Latunde-Dada's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Abas H. Laftah

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chris D. Vulpe

University of California

View shared research outputs
Top Co-Authors

Avatar

Gregory J. Anderson

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge